1
|
Alam A, Smith SC, Gobalakrishnan S, McGinn M, Yakovlev VA, Rabender CS. Uncoupled nitric oxide synthase activity promotes colorectal cancer progression. Front Oncol 2023; 13:1165326. [PMID: 36998441 PMCID: PMC10046306 DOI: 10.3389/fonc.2023.1165326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Increased levels of reactive oxygen/nitrogen species are one hallmark of chronic inflammation contributing to the activation of pro-inflammatory/proliferative pathways. In the cancers analyzed, the tetrahydrobiopterin:dihydrobiopterin ratio is lower than that of the corresponding normal tissue, leading to an uncoupled nitric oxide synthase activity and increased generation of reactive oxygen/nitrogen species. Previously, we demonstrated that prophylactic treatment with sepiapterin, a salvage pathway precursor of tetrahydrobiopterin, prevents dextran sodium sulfate-induced colitis in mice and associated azoxymethane-induced colorectal cancer. Herein, we report that increasing the tetrahydrobiopterin:dihydrobiopterin ratio and recoupling nitric oxide synthase with sepiapterin in the colon cancer cell lines, HCT116 and HT29, inhibit their proliferation and enhance cell death, in part, by Akt/GSK-3β-mediated downregulation of β-catenin. Therapeutic oral gavage with sepiapterin of mice bearing azoxymethane/dextran sodium sulfate-induced colorectal cancer decreased metabolic uptake of [18F]-fluorodeoxyglucose and enhanced apoptosis nine-fold in these tumors. Immunohistochemical analysis of both mouse and human tissues indicated downregulated expression of key enzymes in tetrahydrobiopterin biosynthesis in the colorectal cancer tumors. Human stage 1 colon tumors exhibited a significant decrease in the expression of quinoid dihydropteridine reductase, a key enzyme involved in recycling tetrahydrobiopterin suggesting a potential mechanism for the reduced tetrahydrobiopterin:dihydrobiopterin ratio in these tumors. In summary, sepiapterin treatment of colorectal cancer cells increases the tetrahydrobiopterin:dihydrobiopterin ratio, recouples nitric oxide synthase, and reduces tumor growth. We conclude that nitric oxide synthase coupling may provide a useful therapeutic target for treating patients with colorectal cancer.
Collapse
Affiliation(s)
- Asim Alam
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Steven C. Smith
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Mina McGinn
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Vasily A. Yakovlev
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Christopher S. Rabender
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
2
|
Pillars and Gaps of S-Nitrosylation-Dependent Epigenetic Regulation in Physiology and Cancer. Life (Basel) 2021; 11:life11121424. [PMID: 34947954 PMCID: PMC8704633 DOI: 10.3390/life11121424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Nitric oxide (NO) is a diffusible signaling molecule produced by three isoforms of nitric oxide synthase, which release NO during the metabolism of the amino acid arginine. NO participates in pathophysiological responses of many different tissues, inducing concentration-dependent effect. Indeed, while low NO levels generally have protective effects, higher NO concentrations induce cytotoxic/cytostatic actions. In recent years, evidences have been accumulated unveiling S-nitrosylation as a major NO-dependent post-translational mechanism ruling gene expression. S-nitrosylation is a reversible, highly regulated phenomenon in which NO reacts with one or few specific cysteine residues of target proteins generating S-nitrosothiols. By inducing this chemical modification, NO might exert epigenetic regulation through direct effects on both DNA and histones as well as through indirect actions affecting the functions of transcription factors and transcriptional co-regulators. In this light, S-nitrosylation may also impact on cancer cell gene expression programs. Indeed, it affects different cell pathways and functions ranging from the impairment of DNA damage repair to the modulation of the activity of signal transduction molecules, oncogenes, tumor suppressors, and chromatin remodelers. Nitrosylation is therefore a versatile tool by which NO might control gene expression programs in health and disease.
Collapse
|
3
|
Protein Phosphorylation in Cancer: Role of Nitric Oxide Signaling Pathway. Biomolecules 2021; 11:biom11071009. [PMID: 34356634 PMCID: PMC8301900 DOI: 10.3390/biom11071009] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO), a free radical, plays a critical role in a wide range of physiological and pathological processes. Due to its pleiotropic function, it has been widely investigated in various types of cancers and is strongly associated with cancer development. Mounting pieces of evidence show that NO regulates various cancer-related events, which mainly depends on phosphorylating the key proteins in several signaling pathways. However, phosphorylation of proteins modulated by NO signaling pathway may lead to different effects in different types of cancer, which is complex and remains unclear. Therefore, in this review, we focus on the effect of protein phosphorylation modulated by NO signaling pathway in different types of cancers including breast cancer, lung cancer, prostate cancer, colon cancer, gastric cancer, pancreatic cancer, ovarian cancer, and neuroblastoma. Phosphorylation of key proteins, including p38 MAPK, ERK, PI3K, STAT3, and p53, modified by NO in various signaling pathways affects different cancer-related processes including cell apoptosis, proliferation, angiogenesis, metastasis, and several cancer therapies. Our review links the NO signaling pathway to protein phosphorylation in cancer development and provides new insight into potential targets and cancer therapy.
Collapse
|
4
|
Li M, Liu Y, Deng Y, Pan L, Fu H, Han X, Li Y, Shi H, Wang T. Therapeutic potential of endogenous hydrogen sulfide inhibition in breast cancer (Review). Oncol Rep 2021; 45:68. [PMID: 33760221 PMCID: PMC8020202 DOI: 10.3892/or.2021.8019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Hydrogen sulfide (H2S), the third gas signal molecule, is associated with the modulation of various physiological and pathological processes. Recent studies have reevealed that endogenous H2S may promote proliferation, induce angiogenesis and inhibit apoptosis, thereby stimulating oncogenesis. Conversely, decreased endogenous H2S release suppresses growth of various tumors including breast cancer. This observation suggests an alternative tumor therapy strategy by inhibiting H2S-producing enzymes to reduce the release of endogenous H2S. Breast cancer is the most common type of cancer in women. Due to the lack of approved targeted therapy, its recurrence and metastasis still affect its clinical treatment. In recent years, significant progress has been made in the control of breast cancer by using inhibitors on H2S-producing enzymes. This review summarized the roles of endogenous H2S-producing enzymes in breast cancer and the effects of the enzyme inhibitors on anticancer and anti-metastasis, with the aim of providing new insights for the treatment of breast cancer.
Collapse
Affiliation(s)
- Ming Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Ya Liu
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuying Deng
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Limin Pan
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Han Fu
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Xue Han
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuxi Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Haimei Shi
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Tianxiao Wang
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
5
|
Exploiting S-nitrosylation for cancer therapy: facts and perspectives. Biochem J 2021; 477:3649-3672. [PMID: 33017470 DOI: 10.1042/bcj20200064] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
S-nitrosylation, the post-translational modification of cysteines by nitric oxide, has been implicated in several cellular processes and tissue homeostasis. As a result, alterations in the mechanisms controlling the levels of S-nitrosylated proteins have been found in pathological states. In the last few years, a role in cancer has been proposed, supported by the evidence that various oncoproteins undergo gain- or loss-of-function modifications upon S-nitrosylation. Here, we aim at providing insight into the current knowledge about the role of S-nitrosylation in different aspects of cancer biology and report the main anticancer strategies based on: (i) reducing S-nitrosylation-mediated oncogenic effects, (ii) boosting S-nitrosylation to stimulate cell death, (iii) exploiting S-nitrosylation through synthetic lethality.
Collapse
|
6
|
Khan FH, Dervan E, Bhattacharyya DD, McAuliffe JD, Miranda KM, Glynn SA. The Role of Nitric Oxide in Cancer: Master Regulator or NOt? Int J Mol Sci 2020; 21:ijms21249393. [PMID: 33321789 PMCID: PMC7763974 DOI: 10.3390/ijms21249393] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) is a key player in both the development and suppression of tumourigenesis depending on the source and concentration of NO. In this review, we discuss the mechanisms by which NO induces DNA damage, influences the DNA damage repair response, and subsequently modulates cell cycle arrest. In some circumstances, NO induces cell cycle arrest and apoptosis protecting against tumourigenesis. NO in other scenarios can cause a delay in cell cycle progression, allowing for aberrant DNA repair that promotes the accumulation of mutations and tumour heterogeneity. Within the tumour microenvironment, low to moderate levels of NO derived from tumour and endothelial cells can activate angiogenesis and epithelial-to-mesenchymal transition, promoting an aggressive phenotype. In contrast, high levels of NO derived from inducible nitric oxide synthase (iNOS) expressing M1 and Th1 polarised macrophages and lymphocytes may exert an anti-tumour effect protecting against cancer. It is important to note that the existing evidence on immunomodulation is mainly based on murine iNOS studies which produce higher fluxes of NO than human iNOS. Finally, we discuss different strategies to target NO related pathways therapeutically. Collectively, we present a picture of NO as a master regulator of cancer development and progression.
Collapse
Affiliation(s)
- Faizan H. Khan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Eoin Dervan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Dibyangana D. Bhattacharyya
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Jake D. McAuliffe
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Katrina M. Miranda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA;
| | - Sharon A. Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
- Correspondence:
| |
Collapse
|
7
|
Shi T, Fujita K, Gong J, Nakahara M, Iwama H, Liu S, Yoneyama H, Morishita A, Nomura T, Tani J, Takuma K, Tadokoro T, Himoto T, Oura K, Tsutsui K, Kobara H, Masaki T. Aspirin inhibits hepatocellular carcinoma cell proliferation in vitro and in vivo via inducing cell cycle arrest and apoptosis. Oncol Rep 2020; 44:457-468. [PMID: 32627038 PMCID: PMC7336451 DOI: 10.3892/or.2020.7630] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/10/2020] [Indexed: 12/24/2022] Open
Abstract
Aspirin, a nonsteroidal anti‑inflammatory drug (NSAID), is known to inhibit cell proliferation in a variety of cancers. However, the underlying mechanism of this inhibition remains unknown. We investigated the effects of aspirin on hepatocellular carcinoma (HCC) cells using in vitro and in vivo models. Six HCC cell lines and a liver cancer cell line including Huh‑7 were used in assays that evaluated cell proliferation, cell cycle, and apoptosis. Flow cytometry, enzyme‑linked immunosorbent assay (ELISA), western blot analysis, and phosphorylated receptor tyrosine kinase array were used to evaluate the effects of aspirin on the cells, and microRNAs (miRNAs) were analyzed by a miRNA array chip. The results were validated in vivo using a nude mouse model of Huh‑7‑xenografted tumors. Our results showed that aspirin exhibited an antiproliferative effect on all cell lines. Moreover, aspirin induced G0/G1 cell cycle arrest and modulated the levels of cell cycle‑related molecules such as cyclin E, cyclin D1, and cyclin‑dependent kinase 2 (Cdk2). In addition, aspirin upregulated the levels of caspase‑cleaved cytokeratin 18, increased the proportion of early apoptotic cells, decreased the levels of clusterin and heat shock protein 70 (HSP 70), upregulated the levels of miRNA‑137 and inhibited epidermal growth factor receptor (EGFR) activation. In addition, we observed that aspirin suppressed cell proliferation partially through the miRNA‑137/EGFR pathway. Our in vivo results showed that aspirin reduced the growth of xenograft tumors in nude mice. In conclusion, aspirin was able to inhibit the growth of HCC cells by cell cycle arrest, apoptosis, and alteration of miRNA levels in in vitro and in vivo models.
Collapse
Affiliation(s)
- Tingting Shi
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Koji Fujita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Jian Gong
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Mai Nakahara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Hisakazu Iwama
- Life Science Research Center, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Shi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hirohito Yoneyama
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Takako Nomura
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Kei Takuma
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Tomoko Tadokoro
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Takashi Himoto
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Kyoko Oura
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Kunihiko Tsutsui
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kida, Kagawa 761‑0793, Japan
| |
Collapse
|
8
|
Panettieri S, Paddibhatla I, Chou J, Rajwani R, Moore RS, Goncharuk T, John G, Govind S. Discovery of aspirin-triggered eicosanoid-like mediators in a Drosophila metainflammation blood tumor model. J Cell Sci 2019; 133:jcs.236141. [PMID: 31562189 DOI: 10.1242/jcs.236141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/20/2019] [Indexed: 12/18/2022] Open
Abstract
Epidemiologic studies have linked the use of aspirin to a decline in chronic inflammation that underlies many human diseases, including some cancers. Aspirin reduces the levels of cyclooxygenase-mediated pro-inflammatory prostaglandins, promotes the production of pro-resolution molecules, and triggers the production of anti-inflammatory electrophilic mono-oxygenated (EFOX) lipid mediators. We investigated the effects of aspirin in fruit fly models of chronic inflammation. Ectopic Toll/NF-κB and JAK/STAT signaling in mutant D. melanogaster results in overproliferation of hematopoietic blood progenitors resulting in the formation of granuloma-like tumors. Ectopic JAK-STAT signaling also leads to metabolic inflammation. We report that aspirin-treated mutant flies experience reduction in metabolic inflammation, mitosis, ectopic immune signaling, and macrophage infiltration. Moreover, these flies synthesize 13-HODE, and aspirin triggers 13-oxoODE (13-EFOX-L2) production. Providing the precursor of 13-HODE, linoleic acid, or performing targeted knockdown of the transcription factor STAT in inflammatory blood cells, boosts 13-EFOX-L2 levels while decreasing metabolic inflammation. Thus, hematopoietic cells regulate metabolic inflammation in flies, and their effects can be reversed by pharmaceutical or dietary intervention, suggesting deep phylogenetic conservation in the ability of animals to resolve inflammation and repair tissue damage. These findings can help identify novel treatment targets in humans.
Collapse
Affiliation(s)
- Silvio Panettieri
- Department of Chemistry & Biochemistry, The City College of New York, New York, NY 10031, USA.,PhD Program in Chemistry, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Indira Paddibhatla
- PhD Program in Biology, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA.,Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Jennifer Chou
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Roma Rajwani
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Rebecca S Moore
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Tamara Goncharuk
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - George John
- Department of Chemistry & Biochemistry, The City College of New York, New York, NY 10031, USA .,PhD Program in Chemistry, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Shubha Govind
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA .,PhD Programs in Biology & Biochemistry, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
9
|
Abd El-Hafeez AA, Khalifa HO, Mahdy EAM, Sharma V, Hosoi T, Ghosh P, Ozawa K, Montano MM, Fujimura T, Ibrahim ARN, Abdelhamid MAA, Pack SP, Shouman SA, Kawamoto S. Anticancer effect of nor-wogonin (5, 7, 8-trihydroxyflavone) on human triple-negative breast cancer cells via downregulation of TAK1, NF-κB, and STAT3. Pharmacol Rep 2019; 71:289-298. [PMID: 30826569 DOI: 10.1016/j.pharep.2019.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/26/2018] [Accepted: 01/03/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Nor-wogonin, a polyhydroxy flavone, has been shown to possess antitumor activity. However, the mechanisms responsible for its antitumor activity are poorly studied. Herein, we investigated the mechanisms of nor-wogonin actions in triple-negative breast cancer (TNBC) cells. METHODS Effects of nor-wogonin on cell proliferation and viability of four TNBC cell lines (MDA-MB-231, BT-549, HCC70, and HCC1806) and two non-tumorigenic breast cell lines (MCF-10A and AG11132) were assessed by BrdU incorporation assays and trypan blue dye exclusion tests. Cell cycle and apoptosis analyses were carried out by flow cytometry. Protein expression was analyzed by immunoblotting. RESULTS Nor-wogonin significantly inhibited the growth and decreased the viability of TNBC cells; however, it exhibited no or minimal effects in non-tumorigenic breast cells. Nor-wogonin (40 μM) was a more potent anti-proliferative and cytotoxic agent than wogonin (100 μM) and wogonoside (100 μM), which are structurally related to nor-wogonin. The antitumor effects of nor-wogonin can be attributed to cell cycle arrest via reduction of the expression of cyclin D1, cyclin B1, and CDK1. Furthermore, nor-wogonin induced mitochondrial apoptosis, (as evidenced by the increase in % of cells that are apoptotic), decreases in the mitochondrial membrane potential (ΔΨm), increases in Bax/Bcl-2 ratio, and caspase-3 cleavage. Moreover, nor-wogonin attenuated the expression of the nuclear factor kappa-B and activation of signal transducer and activator of transcription 3 pathways, which can be correlated with suppression of transforming growth factor-β-activated kinase 1 in TNBC cells. CONCLUSION These results showed that nor-wogonin might be a potential multi-target agent for TNBC treatment.
Collapse
Affiliation(s)
- Amer Ali Abd El-Hafeez
- Pharmacotherapy Department, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Global Career Design Center, Hiroshima University, Hiroshima, Japan; Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Hazim O Khalifa
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt; Department of Infectious Diseases, Graduate School of Medicine, International University of Health and Welfare, Narita, Japan
| | | | - Vikas Sharma
- Pharmacology Department, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Toru Hosoi
- Pharmacotherapy Department, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Pradipta Ghosh
- Department of Medicine, University of California, San Diego, La Jolla, CA,USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Koichiro Ozawa
- Pharmacotherapy Department, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Monica M Montano
- Pharmacology Department, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Takashi Fujimura
- Hiroshima Research Center for Healthy Aging (HiHA), Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, Hiroshima, Japan
| | - Ahmed R N Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Mohamed A A Abdelhamid
- Department of Botany and Microbiology, Faculty of Science, Minia University, Minia Egypt; Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea
| | - Samia A Shouman
- Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Seiji Kawamoto
- Hiroshima Research Center for Healthy Aging (HiHA), Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
10
|
Márton M, Tihanyi N, Gyulavári P, Bánhegyi G, Kapuy O. NRF2-regulated cell cycle arrest at early stage of oxidative stress response mechanism. PLoS One 2018; 13:e0207949. [PMID: 30485363 PMCID: PMC6261604 DOI: 10.1371/journal.pone.0207949] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/08/2018] [Indexed: 01/02/2023] Open
Abstract
Oxidative stress results in activation of several signal transduction pathways controlled by the PERK-substrate NRF2 (nuclear factor erythroid 2-related factor 2); meanwhile the ongoing cell division cycle has to be blocked. It has been recently shown that Cyclin D1 got immediately down-regulated via PERK pathway in response to oxidative stress leading to cell cycle arrest. However, the effect of NRF2 on cell cycle regulation has not been explored yet. We aimed to reveal a crosstalk between PERK-substrate NRF2 and the key elements of cell cycle regulatory network upon oxidative stress using molecular biological techniques- Although Cyclin D1 level remained constant, its activity was blocked by various stoichiometric inhibitors (such as p15, p21 and p27) even at low level of oxidative stress. The activity of these CDK inhibitors completely disappeared, when the addition of oxidative agent was combined with silencing of either PERK or NRF2.This further confirms the important role of NRF2 in blocking Cyclin D1 with stoichiometric inhibitors at early stage of oxidative stress.
Collapse
Affiliation(s)
- Margita Márton
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Nikolett Tihanyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Pál Gyulavári
- MTA-SE Pathobiochemistry Research Group, Budapest, Hungary
| | - Gábor Bánhegyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Budapest, Hungary
| | - Orsolya Kapuy
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
11
|
Fan W, Li J, Chen J, Zhu L, Wang Y, Sun B, Hua B, Guo C, Yan Z. Aspirin inhibits the proliferation of synovium-derived mesenchymal stem cells by arresting the cell cycle in the G0/G1 phase. Am J Transl Res 2017; 9:5056-5062. [PMID: 29218104 PMCID: PMC5714790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 11/01/2017] [Indexed: 06/07/2023]
Abstract
Mesenchymal stem cells (MSCs) provide promising applications for clinical treatments. However, patients often take medications that affect the viability of transplanted MSCs. The aim of this study was to assess the effects and underlying mechanism of action of aspirin on the proliferation of MSCs. We showed that aspirin inhibited the growth of MSCs in a concentration- and time-dependent manner. Analysis of cell-cycle distributions showed significantly increased cell populations in the G0/G1 phase and decreased cell populations in the S phase and G2/M phase with increasing concentrations of aspirin. We further analyzed the expression of cyclins and found that the level of cyclin D1 was significantly reduced after aspirin treatment, while there was no obvious effect on the levels of cyclin A2 and cyclin E1. Because we showed that the expression of miRNA145 was significantly increased after aspirin treatment, we further transfected MSCs with an miRNA145 mimic or miRNA145 inhibitor. Transfection with the miRNA145 mimic resulted in decreased expression of cyclin D1, while transfection with miRNA145 inhibitor resulted in increased expression of cyclin D1. Transfection with miRNA145 inhibitor abolished the downregulation of cyclin D induced by aspirin. The results suggested that aspirin inhibited the proliferation of MSCs and caused cell-cycle arrest in the G0/G1 phase through downregulation of cyclin D1, which could be related to the increased expression of miRNA145.
Collapse
Affiliation(s)
- Wenshuai Fan
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Jinghuan Li
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Jifei Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Liang Zhu
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Yiming Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Bolin Sun
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Bingxuan Hua
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Changan Guo
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
12
|
Liao Q, Huang YM, Fan W, Li C, Yang H. Endothelial nitric oxide synthase deficiency influences normal cell cycle progression and apoptosis in trabecular meshwork cells. Int J Ophthalmol 2016; 9:799-803. [PMID: 27366677 DOI: 10.18240/ijo.2016.06.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/27/2015] [Indexed: 11/23/2022] Open
Abstract
AIM To clarify how the endothelial nitric oxide synthase (eNOS, NOS3) make effect on outflow facility through the trabecular meshwork (TM). METHODS Inhibition of NOS3 gene expression in human TM cells were conducted by three siRNAs. Then the mRNA and protein levels of NOS3 in siRNA-treated and negative control (NC) cells were determined, still were the collagen, type IV, alpha 1 (COL4A1) and fibronectin 1 by real-time PCR and Western blot analysis. In addition, NOS3 concentrations in culture supernatant fluids of TM cells were measured. Cell cycle and cell apoptosis analysis were performed using flow cytometry. RESULTS The mRNA level of NOS3 was decreased by three different siRNA interference, similar results were obtained not only of the relative levels of NOS3 protein, but also the expression levels of COL4A1 and fibronectin 1. The number of cells in S phase was decreased, while contrary result was obtained in G2 phase. The number of apoptotic cells in siRNA-treated groups were significant increased compared to the NC samples. CONCLUSION Abnormal NOS3 expression can make effects on the proteins levels of extracellular matrix component (e.g. fibronectin 1 and COL4A1). Reduced NOS3 restrains the TM cell cycle progression at the G2/M-phase transition and induced cell apoptosis.
Collapse
Affiliation(s)
- Qiong Liao
- Department of Ophthalmology, Xinqiao Hospital of Third Military Medical University, Chongqing 400016, China
| | - Yan-Ming Huang
- Department of Ophthalmology, Xinqiao Hospital of Third Military Medical University, Chongqing 400016, China
| | - Wei Fan
- Department of Ophthalmology, Xinqiao Hospital of Third Military Medical University, Chongqing 400016, China
| | - Chan Li
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University of Chongqing Medical University, Chongqing 400016, China
| | - Hong Yang
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University, Chongqing 400016, China
| |
Collapse
|
13
|
Byrnes KA, Phatak P, Mansour D, Xiao L, Zou T, Rao JN, Turner DJ, Wang JY, Donahue JM. Overexpression of miR-199a-5p decreases esophageal cancer cell proliferation through repression of mitogen-activated protein kinase kinase kinase-11 (MAP3K11). Oncotarget 2016; 7:8756-70. [PMID: 26717044 PMCID: PMC4891002 DOI: 10.18632/oncotarget.6752] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 11/16/2015] [Indexed: 01/23/2023] Open
Abstract
Studies examining the oncogenic or tumor suppressive functions of dysregulated microRNAs (miRs) in cancer cells may also identify novel miR targets, which can themselves serve as therapeutic targets. Using array analysis, we have previously determined that miR-199a-5p was the most downregulated miR in two esophageal cancer cell lines compared to esophageal epithelial cells. MiR-199a-5p is predicted to bind mitogen-activated protein kinase kinase kinase 11 (MAP3K11) mRNA with high affinity. In this study, we observed that MAP3K11 is markedly overexpressed in esophageal cancer cell lines. Forced expression of miR-199a-5p in these cells leads to a decrease in the mRNA and protein levels of MAP3K11, due to decreased MAP3K11 mRNA stability. A direct binding interaction between miR-199a-5p and MAP3K11 mRNA is demonstrated using biotin pull-down assays and heterologous luciferase reporter constructs and confirmed by mutational analysis. Finally, forced expression of miR-199a-5p decreases proliferation of esophageal cancer cells by inducing G2/M arrest. This effect is mediated, in part, by decreased transcription of cyclin D1, due to reduced MAP3K11-mediated phosphorylation of c-Jun. These findings suggest that miR-199a-5p acts as a tumor suppressor in esophageal cancer cells and that its downregulation contributes to enhanced cellular proliferation by targeting MAP3K11.
Collapse
Affiliation(s)
- Kimberly A. Byrnes
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Pornima Phatak
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Daniel Mansour
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Lan Xiao
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Tongtong Zou
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jaladanki N. Rao
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Douglas J. Turner
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jian-Ying Wang
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
| | - James M. Donahue
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| |
Collapse
|
14
|
Nitric Oxide-Releasing Aspirin Suppresses NF-κB Signaling in Estrogen Receptor Negative Breast Cancer Cells in Vitro and in Vivo. Molecules 2015; 20:12481-99. [PMID: 26184135 PMCID: PMC6331965 DOI: 10.3390/molecules200712481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/11/2015] [Accepted: 07/06/2015] [Indexed: 01/29/2023] Open
Abstract
Estrogen receptor negative (ER(-)) breast cancer is aggressive, responds poorly to current treatments and has a poor prognosis. The NF-κB signaling pathway is implicated in ER(-) tumorigenesis. Aspirin (ASA) is chemopreventive against ER(+) but not for ER(-) breast cancers. Nitric oxide-releasing aspirin (NO-ASA) is a safer ASA where ASA is linked to an NO-releasing moiety through a spacer. In vitro, we investigated anti-proliferation effects of NO-ASA (para- and meta-isomers) against ER(-) breast cancer cells MDA-MB-231 and SK-BR-23, effects on NF-κB signaling, and reactive oxygen species by standard techniques. In vivo, effects of NO-ASA were evaluated in a mouse xenograft model using MDA-MB-231 cells. p-NO-ASA inhibited the growth of MDA-MB-231 and SK-BR-3 cells at 24 h, the respective IC50s were 13 ± 2 and 17 ± 2 μM; ASA had an IC50 of >3000 μM in both cell lines. The IC50s for m-NO-ASA in MDA-MB-231 and SK-BR-3 were 173 ± 15 and 185 ± 12 μM, respectively, therefore, implying p-NO-ASA as a stronger inhibitor of growth p-NO-ASA reduced cell growth by inhibiting proliferation, inducing apoptosis and causing G0/G1 cell cycle block. Activation of NF-κB was inhibited by both isomers as demonstrated by decreases in NF-κB-DNA binding and luciferase activity at 24 h, However, m-NO-ASA produced transient effects at 3 h such as increased NF-κB-DNA-binding, increased levels of nuclear p50, even though both isomers inhibited IκB degradation. Increase in nuclear p50 by m-NO-ASA was associated with translocation of p50 in to the nucleus as observed by immunoflouresence at 3 h. NO-ASA induced reactive oxygen species (ROS) as evidenced by overall increases in both H2DCFDA (2',7'-dichlorodihydrofluorescein) and DHE (dihydroethidium)-derived fluorescence. Inhibition of ROS by N-acetyl-cysteine reversed the m-NO-ASA-mediated translocation of p50 in to the nucleus. In xenografts, p-NO-ASA inhibited tumor growth by inhibiting proliferation (PCNA and tumor volume), inducing apoptosis (TUNEL positive cells) and reducing NF-κB expression. Both isomers inhibit cancer cells, inhibit NF-κB pathway and induce ROS, and have potential as anticancer compounds.
Collapse
|
15
|
McGoldrick CA, Jiang YL, Brannon M, Krishnan K, Stone WL. In vitro evaluation of novel N-acetylalaninate prodrugs that selectively induce apoptosis in prostate cancer cells. BMC Cancer 2014; 14:675. [PMID: 25234292 PMCID: PMC4180535 DOI: 10.1186/1471-2407-14-675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 09/16/2014] [Indexed: 12/05/2022] Open
Abstract
Background Cancer cell esterases are often overexpressed and can have chiral specificities different from that of the corresponding normal cells and can, therefore, be useful targets for activating chemotherapeutic prodrug esters. Prodrug esters are inactive compounds that can be preferentially activated by esterase enzymes. Moreover, cancer cells often exhibit a high level of intrinsic oxidative stress due to an increased formation of reactive oxygen species (ROS) and a decreased expression of some enzymatic antioxidants. Prodrugs designed to induce additional oxidative stress can selectively induce apoptosis in cancer cells already exhibiting a high level of intrinsic oxidative stress. This study focused on the in vitro evaluation of four novel prodrug esters: the R- and S- chiral esters of 4-[(nitrooxy)methyl]phenyl N-acetylalaninate (R- and S-NPAA) and the R- and S- chiral esters of 4-[(nitrooxy)methyl]naphth-1-yl N-acetylalaninate (R- and S-NQM), which are activated, to varying extents, by oxidized protein hydrolase (OPH, EC 3.4.19.1) yielding a quinone methide (QM) intermediate capable of depleting glutathione (GSH), a key intracellular antioxidant. OPH is a serine esterase/protease that is overexpressed in some human tumors and cancer cell lines. Methods To evaluate the chiral ester prodrugs, we monitored cellular GSH depletion, cellular protein carbonyl levels (an oxidative stress biomarker) and cell viability in tumorigenic and nontumorigenic prostate cancer cell lines. Results We found that the prodrugs were activated by OPH and subsequently depleted GSH. The S-chiral ester of NPAA (S-NPAA) was two-fold more effective than the R-chiral ester (R-NPAA) in depleting GSH, increasing oxidative stress, inducing apoptosis, and decreasing cell viability in tumorigenic prostate LNCaP cells but had little effect on non-tumorigenic RWPE-1 cells. In addition, we found that that S-NPAA induced apoptosis and decreased cell viability in tumorigenic DU145 and PC3 prostate cell lines. Similar results were found in a COS-7 model that overexpressed active human OPH (COS-7-OPH). Conclusions Our results suggest that prostate tumors overexpressing OPH and/or exhibiting a high level of intrinsic oxidative stress may be susceptible to QM generating prodrug esters that are targeted to OPH with little effect on non-tumorigenic prostate cells.
Collapse
Affiliation(s)
| | | | | | | | - William L Stone
- Department of Pediatrics, East Tennessee State University, Johnson City, TN 37614-0578, USA.
| |
Collapse
|
16
|
Xu XY, Song GQ, Yu YQ, Ma HY, Ma L, Jin YN. Apoptosis and G2/M arrest induced by Allium ursinum (ramson) watery extract in an AGS gastric cancer cell line. Onco Targets Ther 2013; 6:779-83. [PMID: 23836991 PMCID: PMC3699351 DOI: 10.2147/ott.s45865] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The present study was designed to determine whether Allium ursinum L (ramson) could inhibit the proliferation of human AGS gastric cancer cells. Furthermore, we attempted to determine whether this inhibition could occur by targeting regulatory elements of the cell cycle. Methods Flow cytometry was used to observe apoptosis and the cell cycle in AGS cell lines treated or not treated with ramson watery extract. Proteins related to the cell cycle were detected by Western blotting. Caspase activity was measured using a colorimetric assay kit according to the manufacturer’s instructions. Results Ramson watery extract induced apoptosis and G2/M phase arrest in AGS cells. Western blotting showed that cyclin B was inhibited by ramson watery extract. However, G1 phase-related proteins remain unchanged after treatment. Conclusion Our results indicate that ramson effectively sup pressed proliferation and induced apoptosis and G2/M arrest in AGS cells by regulating elements of the cell cycle.
Collapse
Affiliation(s)
- Xiao-Yan Xu
- Department of Pathophysiology, Institute of Pathology and Pathophysiology, College of Basic Medicine, China Medical University
| | | | | | | | | | | |
Collapse
|